51
|
Panzeri S, Zanella S, Arosio D, Vahdati L, Dal Corso A, Pignataro L, Paolillo M, Schinelli S, Belvisi L, Gennari C, Piarulli U. Cyclic isoDGR and RGD peptidomimetics containing bifunctional diketopiperazine scaffolds are integrin antagonists. Chemistry 2015; 21:6265-71. [PMID: 25761230 DOI: 10.1002/chem.201406567] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Indexed: 01/18/2023]
Abstract
The cyclo[DKP-isoDGR] peptidomimetics 2-5, containing bifunctional diketopiperazine (DKP) scaffolds that differ in the configuration of the two DKP stereocenters and in the substitution at the DKP nitrogen atoms, were prepared and examined in vitro in competitive binding assays with purified αv β3 and αv β5 integrin receptors. IC50 values ranged from low nanomolar (ligand 3) to submicromolar with αv β3 integrin. The biological activities of ligands cyclo[DKP3-RGD] 1 and cyclo[DKP3-isoDGR] 3, bearing the same bifunctional DKP scaffold and showing similar αV β3 integrin binding values, were compared in terms of their cellular effects in human U373 glioblastoma cells. Compounds 1 and 3 displayed overlapping inhibitory effects on the FAK/Akt integrin activated transduction pathway and on integrin-mediated cell infiltration processes, and qualify therefore, despite the different RGD and isoDGR sequences, as integrin antagonists. Both compounds induced apoptosis in glioma cells after 72 hour treatment.
Collapse
Affiliation(s)
- Silvia Panzeri
- Università degli Studi dell'Insubria, Dipartimento di Scienza e Alta Tecnologia, Via Valleggio 11, 22100 Como (Italy)
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
52
|
Nabors LB, Fink KL, Mikkelsen T, Grujicic D, Tarnawski R, Nam DH, Mazurkiewicz M, Salacz M, Ashby L, Zagonel V, Depenni R, Perry JR, Hicking C, Picard M, Hegi ME, Lhermitte B, Reardon DA. Two cilengitide regimens in combination with standard treatment for patients with newly diagnosed glioblastoma and unmethylated MGMT gene promoter: results of the open-label, controlled, randomized phase II CORE study. Neuro Oncol 2015; 17:708-17. [PMID: 25762461 DOI: 10.1093/neuonc/nou356] [Citation(s) in RCA: 178] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 12/12/2014] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Survival outcomes for patients with glioblastoma remain poor, particularly for patients with unmethylated O(6)-methylguanine-DNA methyltransferase (MGMT) gene promoter. This phase II, randomized, open-label, multicenter trial investigated the efficacy and safety of 2 dose regimens of the selective integrin inhibitor cilengitide combined with standard chemoradiotherapy in patients with newly diagnosed glioblastoma and an unmethylated MGMT promoter. METHODS Overall, 265 patients were randomized (1:1:1) to standard cilengitide (2000 mg 2×/wk; n = 88), intensive cilengitide (2000 mg 5×/wk during wk 1-6, thereafter 2×/wk; n = 88), or a control arm (chemoradiotherapy alone; n = 89). Cilengitide was administered intravenously in combination with daily temozolomide (TMZ) and concomitant radiotherapy (RT; wk 1-6), followed by TMZ maintenance therapy (TMZ/RT→TMZ). The primary endpoint was overall survival; secondary endpoints included progression-free survival, pharmacokinetics, and safety and tolerability. RESULTS Median overall survival was 16.3 months in the standard cilengitide arm (hazard ratio [HR], 0.686; 95% CI: 0.484, 0.972; P = .032) and 14.5 months in the intensive cilengitide arm (HR, 0.858; 95% CI: 0.612, 1.204; P = .3771) versus 13.4 months in the control arm. Median progression-free survival assessed per independent review committee was 5.6 months (HR, 0.822; 95% CI: 0.595, 1.134) and 5.9 months (HR, 0.794; 95% CI: 0.575, 1.096) in the standard and intensive cilengitide arms, respectively, versus 4.1 months in the control arm. Cilengitide was well tolerated. CONCLUSIONS Standard and intensive cilengitide dose regimens were well tolerated in combination with TMZ/RT→TMZ. Inconsistent overall survival and progression-free survival outcomes and a limited sample size did not allow firm conclusions regarding clinical efficacy in this exploratory phase II study.
Collapse
Affiliation(s)
- L Burt Nabors
- University of Alabama at Birmingham, Birmingham, Alabama (L.B.N.); Baylor University Medical Center, Dallas, Texas (K.L.F.); Henry Ford Hospital, Detroit, Michigan (T.M.); Clinic for Neurosurgery, Clinical Center of Serbia, Medical Faculty University of Belgrade, Belgrade, Serbia (D.G.); Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology Gliwice Branch, Radiotherapy and Chemotherapy Clinic, Gliwice, Poland (R.T.); Samsung Medical Center, Seoul, South Korea (D.H.N.); Centrum Onkologii Ziemi Lubelskiej, Lublin, Poland (M.M.); St. Luke's Brain Tumor Center, St. Luke's Hospital, Kansas City, Missouri (M.S.); Barrow Neurological Institute, Phoenix, Arizona (L.A.); Medical Oncology Unit 1, IOV, IRCCS, Padova, Italy (V.Z.); Policlinico di Modena, Modena, Italy (R.D.); Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (J.R.P.); Merck KGaA, Darmstadt, Germany (C.H., M.P.); Department of Clinical Neurosciences, University Hospital Lausanne, Lausanne, Switzerland (M.E.H.); Institute of Pathology, University Hospital Lausanne, Lausanne, Switzerland (B.L.); Dana-Farber Cancer Institute, Boston, Massachusetts (D.A.R.)
| | - Karen L Fink
- University of Alabama at Birmingham, Birmingham, Alabama (L.B.N.); Baylor University Medical Center, Dallas, Texas (K.L.F.); Henry Ford Hospital, Detroit, Michigan (T.M.); Clinic for Neurosurgery, Clinical Center of Serbia, Medical Faculty University of Belgrade, Belgrade, Serbia (D.G.); Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology Gliwice Branch, Radiotherapy and Chemotherapy Clinic, Gliwice, Poland (R.T.); Samsung Medical Center, Seoul, South Korea (D.H.N.); Centrum Onkologii Ziemi Lubelskiej, Lublin, Poland (M.M.); St. Luke's Brain Tumor Center, St. Luke's Hospital, Kansas City, Missouri (M.S.); Barrow Neurological Institute, Phoenix, Arizona (L.A.); Medical Oncology Unit 1, IOV, IRCCS, Padova, Italy (V.Z.); Policlinico di Modena, Modena, Italy (R.D.); Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (J.R.P.); Merck KGaA, Darmstadt, Germany (C.H., M.P.); Department of Clinical Neurosciences, University Hospital Lausanne, Lausanne, Switzerland (M.E.H.); Institute of Pathology, University Hospital Lausanne, Lausanne, Switzerland (B.L.); Dana-Farber Cancer Institute, Boston, Massachusetts (D.A.R.)
| | - Tom Mikkelsen
- University of Alabama at Birmingham, Birmingham, Alabama (L.B.N.); Baylor University Medical Center, Dallas, Texas (K.L.F.); Henry Ford Hospital, Detroit, Michigan (T.M.); Clinic for Neurosurgery, Clinical Center of Serbia, Medical Faculty University of Belgrade, Belgrade, Serbia (D.G.); Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology Gliwice Branch, Radiotherapy and Chemotherapy Clinic, Gliwice, Poland (R.T.); Samsung Medical Center, Seoul, South Korea (D.H.N.); Centrum Onkologii Ziemi Lubelskiej, Lublin, Poland (M.M.); St. Luke's Brain Tumor Center, St. Luke's Hospital, Kansas City, Missouri (M.S.); Barrow Neurological Institute, Phoenix, Arizona (L.A.); Medical Oncology Unit 1, IOV, IRCCS, Padova, Italy (V.Z.); Policlinico di Modena, Modena, Italy (R.D.); Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (J.R.P.); Merck KGaA, Darmstadt, Germany (C.H., M.P.); Department of Clinical Neurosciences, University Hospital Lausanne, Lausanne, Switzerland (M.E.H.); Institute of Pathology, University Hospital Lausanne, Lausanne, Switzerland (B.L.); Dana-Farber Cancer Institute, Boston, Massachusetts (D.A.R.)
| | - Danica Grujicic
- University of Alabama at Birmingham, Birmingham, Alabama (L.B.N.); Baylor University Medical Center, Dallas, Texas (K.L.F.); Henry Ford Hospital, Detroit, Michigan (T.M.); Clinic for Neurosurgery, Clinical Center of Serbia, Medical Faculty University of Belgrade, Belgrade, Serbia (D.G.); Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology Gliwice Branch, Radiotherapy and Chemotherapy Clinic, Gliwice, Poland (R.T.); Samsung Medical Center, Seoul, South Korea (D.H.N.); Centrum Onkologii Ziemi Lubelskiej, Lublin, Poland (M.M.); St. Luke's Brain Tumor Center, St. Luke's Hospital, Kansas City, Missouri (M.S.); Barrow Neurological Institute, Phoenix, Arizona (L.A.); Medical Oncology Unit 1, IOV, IRCCS, Padova, Italy (V.Z.); Policlinico di Modena, Modena, Italy (R.D.); Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (J.R.P.); Merck KGaA, Darmstadt, Germany (C.H., M.P.); Department of Clinical Neurosciences, University Hospital Lausanne, Lausanne, Switzerland (M.E.H.); Institute of Pathology, University Hospital Lausanne, Lausanne, Switzerland (B.L.); Dana-Farber Cancer Institute, Boston, Massachusetts (D.A.R.)
| | - Rafal Tarnawski
- University of Alabama at Birmingham, Birmingham, Alabama (L.B.N.); Baylor University Medical Center, Dallas, Texas (K.L.F.); Henry Ford Hospital, Detroit, Michigan (T.M.); Clinic for Neurosurgery, Clinical Center of Serbia, Medical Faculty University of Belgrade, Belgrade, Serbia (D.G.); Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology Gliwice Branch, Radiotherapy and Chemotherapy Clinic, Gliwice, Poland (R.T.); Samsung Medical Center, Seoul, South Korea (D.H.N.); Centrum Onkologii Ziemi Lubelskiej, Lublin, Poland (M.M.); St. Luke's Brain Tumor Center, St. Luke's Hospital, Kansas City, Missouri (M.S.); Barrow Neurological Institute, Phoenix, Arizona (L.A.); Medical Oncology Unit 1, IOV, IRCCS, Padova, Italy (V.Z.); Policlinico di Modena, Modena, Italy (R.D.); Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (J.R.P.); Merck KGaA, Darmstadt, Germany (C.H., M.P.); Department of Clinical Neurosciences, University Hospital Lausanne, Lausanne, Switzerland (M.E.H.); Institute of Pathology, University Hospital Lausanne, Lausanne, Switzerland (B.L.); Dana-Farber Cancer Institute, Boston, Massachusetts (D.A.R.)
| | - Do Hyun Nam
- University of Alabama at Birmingham, Birmingham, Alabama (L.B.N.); Baylor University Medical Center, Dallas, Texas (K.L.F.); Henry Ford Hospital, Detroit, Michigan (T.M.); Clinic for Neurosurgery, Clinical Center of Serbia, Medical Faculty University of Belgrade, Belgrade, Serbia (D.G.); Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology Gliwice Branch, Radiotherapy and Chemotherapy Clinic, Gliwice, Poland (R.T.); Samsung Medical Center, Seoul, South Korea (D.H.N.); Centrum Onkologii Ziemi Lubelskiej, Lublin, Poland (M.M.); St. Luke's Brain Tumor Center, St. Luke's Hospital, Kansas City, Missouri (M.S.); Barrow Neurological Institute, Phoenix, Arizona (L.A.); Medical Oncology Unit 1, IOV, IRCCS, Padova, Italy (V.Z.); Policlinico di Modena, Modena, Italy (R.D.); Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (J.R.P.); Merck KGaA, Darmstadt, Germany (C.H., M.P.); Department of Clinical Neurosciences, University Hospital Lausanne, Lausanne, Switzerland (M.E.H.); Institute of Pathology, University Hospital Lausanne, Lausanne, Switzerland (B.L.); Dana-Farber Cancer Institute, Boston, Massachusetts (D.A.R.)
| | - Maria Mazurkiewicz
- University of Alabama at Birmingham, Birmingham, Alabama (L.B.N.); Baylor University Medical Center, Dallas, Texas (K.L.F.); Henry Ford Hospital, Detroit, Michigan (T.M.); Clinic for Neurosurgery, Clinical Center of Serbia, Medical Faculty University of Belgrade, Belgrade, Serbia (D.G.); Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology Gliwice Branch, Radiotherapy and Chemotherapy Clinic, Gliwice, Poland (R.T.); Samsung Medical Center, Seoul, South Korea (D.H.N.); Centrum Onkologii Ziemi Lubelskiej, Lublin, Poland (M.M.); St. Luke's Brain Tumor Center, St. Luke's Hospital, Kansas City, Missouri (M.S.); Barrow Neurological Institute, Phoenix, Arizona (L.A.); Medical Oncology Unit 1, IOV, IRCCS, Padova, Italy (V.Z.); Policlinico di Modena, Modena, Italy (R.D.); Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (J.R.P.); Merck KGaA, Darmstadt, Germany (C.H., M.P.); Department of Clinical Neurosciences, University Hospital Lausanne, Lausanne, Switzerland (M.E.H.); Institute of Pathology, University Hospital Lausanne, Lausanne, Switzerland (B.L.); Dana-Farber Cancer Institute, Boston, Massachusetts (D.A.R.)
| | - Michael Salacz
- University of Alabama at Birmingham, Birmingham, Alabama (L.B.N.); Baylor University Medical Center, Dallas, Texas (K.L.F.); Henry Ford Hospital, Detroit, Michigan (T.M.); Clinic for Neurosurgery, Clinical Center of Serbia, Medical Faculty University of Belgrade, Belgrade, Serbia (D.G.); Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology Gliwice Branch, Radiotherapy and Chemotherapy Clinic, Gliwice, Poland (R.T.); Samsung Medical Center, Seoul, South Korea (D.H.N.); Centrum Onkologii Ziemi Lubelskiej, Lublin, Poland (M.M.); St. Luke's Brain Tumor Center, St. Luke's Hospital, Kansas City, Missouri (M.S.); Barrow Neurological Institute, Phoenix, Arizona (L.A.); Medical Oncology Unit 1, IOV, IRCCS, Padova, Italy (V.Z.); Policlinico di Modena, Modena, Italy (R.D.); Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (J.R.P.); Merck KGaA, Darmstadt, Germany (C.H., M.P.); Department of Clinical Neurosciences, University Hospital Lausanne, Lausanne, Switzerland (M.E.H.); Institute of Pathology, University Hospital Lausanne, Lausanne, Switzerland (B.L.); Dana-Farber Cancer Institute, Boston, Massachusetts (D.A.R.)
| | - Lynn Ashby
- University of Alabama at Birmingham, Birmingham, Alabama (L.B.N.); Baylor University Medical Center, Dallas, Texas (K.L.F.); Henry Ford Hospital, Detroit, Michigan (T.M.); Clinic for Neurosurgery, Clinical Center of Serbia, Medical Faculty University of Belgrade, Belgrade, Serbia (D.G.); Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology Gliwice Branch, Radiotherapy and Chemotherapy Clinic, Gliwice, Poland (R.T.); Samsung Medical Center, Seoul, South Korea (D.H.N.); Centrum Onkologii Ziemi Lubelskiej, Lublin, Poland (M.M.); St. Luke's Brain Tumor Center, St. Luke's Hospital, Kansas City, Missouri (M.S.); Barrow Neurological Institute, Phoenix, Arizona (L.A.); Medical Oncology Unit 1, IOV, IRCCS, Padova, Italy (V.Z.); Policlinico di Modena, Modena, Italy (R.D.); Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (J.R.P.); Merck KGaA, Darmstadt, Germany (C.H., M.P.); Department of Clinical Neurosciences, University Hospital Lausanne, Lausanne, Switzerland (M.E.H.); Institute of Pathology, University Hospital Lausanne, Lausanne, Switzerland (B.L.); Dana-Farber Cancer Institute, Boston, Massachusetts (D.A.R.)
| | - Vittorina Zagonel
- University of Alabama at Birmingham, Birmingham, Alabama (L.B.N.); Baylor University Medical Center, Dallas, Texas (K.L.F.); Henry Ford Hospital, Detroit, Michigan (T.M.); Clinic for Neurosurgery, Clinical Center of Serbia, Medical Faculty University of Belgrade, Belgrade, Serbia (D.G.); Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology Gliwice Branch, Radiotherapy and Chemotherapy Clinic, Gliwice, Poland (R.T.); Samsung Medical Center, Seoul, South Korea (D.H.N.); Centrum Onkologii Ziemi Lubelskiej, Lublin, Poland (M.M.); St. Luke's Brain Tumor Center, St. Luke's Hospital, Kansas City, Missouri (M.S.); Barrow Neurological Institute, Phoenix, Arizona (L.A.); Medical Oncology Unit 1, IOV, IRCCS, Padova, Italy (V.Z.); Policlinico di Modena, Modena, Italy (R.D.); Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (J.R.P.); Merck KGaA, Darmstadt, Germany (C.H., M.P.); Department of Clinical Neurosciences, University Hospital Lausanne, Lausanne, Switzerland (M.E.H.); Institute of Pathology, University Hospital Lausanne, Lausanne, Switzerland (B.L.); Dana-Farber Cancer Institute, Boston, Massachusetts (D.A.R.)
| | - Roberta Depenni
- University of Alabama at Birmingham, Birmingham, Alabama (L.B.N.); Baylor University Medical Center, Dallas, Texas (K.L.F.); Henry Ford Hospital, Detroit, Michigan (T.M.); Clinic for Neurosurgery, Clinical Center of Serbia, Medical Faculty University of Belgrade, Belgrade, Serbia (D.G.); Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology Gliwice Branch, Radiotherapy and Chemotherapy Clinic, Gliwice, Poland (R.T.); Samsung Medical Center, Seoul, South Korea (D.H.N.); Centrum Onkologii Ziemi Lubelskiej, Lublin, Poland (M.M.); St. Luke's Brain Tumor Center, St. Luke's Hospital, Kansas City, Missouri (M.S.); Barrow Neurological Institute, Phoenix, Arizona (L.A.); Medical Oncology Unit 1, IOV, IRCCS, Padova, Italy (V.Z.); Policlinico di Modena, Modena, Italy (R.D.); Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (J.R.P.); Merck KGaA, Darmstadt, Germany (C.H., M.P.); Department of Clinical Neurosciences, University Hospital Lausanne, Lausanne, Switzerland (M.E.H.); Institute of Pathology, University Hospital Lausanne, Lausanne, Switzerland (B.L.); Dana-Farber Cancer Institute, Boston, Massachusetts (D.A.R.)
| | - James R Perry
- University of Alabama at Birmingham, Birmingham, Alabama (L.B.N.); Baylor University Medical Center, Dallas, Texas (K.L.F.); Henry Ford Hospital, Detroit, Michigan (T.M.); Clinic for Neurosurgery, Clinical Center of Serbia, Medical Faculty University of Belgrade, Belgrade, Serbia (D.G.); Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology Gliwice Branch, Radiotherapy and Chemotherapy Clinic, Gliwice, Poland (R.T.); Samsung Medical Center, Seoul, South Korea (D.H.N.); Centrum Onkologii Ziemi Lubelskiej, Lublin, Poland (M.M.); St. Luke's Brain Tumor Center, St. Luke's Hospital, Kansas City, Missouri (M.S.); Barrow Neurological Institute, Phoenix, Arizona (L.A.); Medical Oncology Unit 1, IOV, IRCCS, Padova, Italy (V.Z.); Policlinico di Modena, Modena, Italy (R.D.); Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (J.R.P.); Merck KGaA, Darmstadt, Germany (C.H., M.P.); Department of Clinical Neurosciences, University Hospital Lausanne, Lausanne, Switzerland (M.E.H.); Institute of Pathology, University Hospital Lausanne, Lausanne, Switzerland (B.L.); Dana-Farber Cancer Institute, Boston, Massachusetts (D.A.R.)
| | - Christine Hicking
- University of Alabama at Birmingham, Birmingham, Alabama (L.B.N.); Baylor University Medical Center, Dallas, Texas (K.L.F.); Henry Ford Hospital, Detroit, Michigan (T.M.); Clinic for Neurosurgery, Clinical Center of Serbia, Medical Faculty University of Belgrade, Belgrade, Serbia (D.G.); Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology Gliwice Branch, Radiotherapy and Chemotherapy Clinic, Gliwice, Poland (R.T.); Samsung Medical Center, Seoul, South Korea (D.H.N.); Centrum Onkologii Ziemi Lubelskiej, Lublin, Poland (M.M.); St. Luke's Brain Tumor Center, St. Luke's Hospital, Kansas City, Missouri (M.S.); Barrow Neurological Institute, Phoenix, Arizona (L.A.); Medical Oncology Unit 1, IOV, IRCCS, Padova, Italy (V.Z.); Policlinico di Modena, Modena, Italy (R.D.); Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (J.R.P.); Merck KGaA, Darmstadt, Germany (C.H., M.P.); Department of Clinical Neurosciences, University Hospital Lausanne, Lausanne, Switzerland (M.E.H.); Institute of Pathology, University Hospital Lausanne, Lausanne, Switzerland (B.L.); Dana-Farber Cancer Institute, Boston, Massachusetts (D.A.R.)
| | - Martin Picard
- University of Alabama at Birmingham, Birmingham, Alabama (L.B.N.); Baylor University Medical Center, Dallas, Texas (K.L.F.); Henry Ford Hospital, Detroit, Michigan (T.M.); Clinic for Neurosurgery, Clinical Center of Serbia, Medical Faculty University of Belgrade, Belgrade, Serbia (D.G.); Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology Gliwice Branch, Radiotherapy and Chemotherapy Clinic, Gliwice, Poland (R.T.); Samsung Medical Center, Seoul, South Korea (D.H.N.); Centrum Onkologii Ziemi Lubelskiej, Lublin, Poland (M.M.); St. Luke's Brain Tumor Center, St. Luke's Hospital, Kansas City, Missouri (M.S.); Barrow Neurological Institute, Phoenix, Arizona (L.A.); Medical Oncology Unit 1, IOV, IRCCS, Padova, Italy (V.Z.); Policlinico di Modena, Modena, Italy (R.D.); Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (J.R.P.); Merck KGaA, Darmstadt, Germany (C.H., M.P.); Department of Clinical Neurosciences, University Hospital Lausanne, Lausanne, Switzerland (M.E.H.); Institute of Pathology, University Hospital Lausanne, Lausanne, Switzerland (B.L.); Dana-Farber Cancer Institute, Boston, Massachusetts (D.A.R.)
| | - Monika E Hegi
- University of Alabama at Birmingham, Birmingham, Alabama (L.B.N.); Baylor University Medical Center, Dallas, Texas (K.L.F.); Henry Ford Hospital, Detroit, Michigan (T.M.); Clinic for Neurosurgery, Clinical Center of Serbia, Medical Faculty University of Belgrade, Belgrade, Serbia (D.G.); Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology Gliwice Branch, Radiotherapy and Chemotherapy Clinic, Gliwice, Poland (R.T.); Samsung Medical Center, Seoul, South Korea (D.H.N.); Centrum Onkologii Ziemi Lubelskiej, Lublin, Poland (M.M.); St. Luke's Brain Tumor Center, St. Luke's Hospital, Kansas City, Missouri (M.S.); Barrow Neurological Institute, Phoenix, Arizona (L.A.); Medical Oncology Unit 1, IOV, IRCCS, Padova, Italy (V.Z.); Policlinico di Modena, Modena, Italy (R.D.); Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (J.R.P.); Merck KGaA, Darmstadt, Germany (C.H., M.P.); Department of Clinical Neurosciences, University Hospital Lausanne, Lausanne, Switzerland (M.E.H.); Institute of Pathology, University Hospital Lausanne, Lausanne, Switzerland (B.L.); Dana-Farber Cancer Institute, Boston, Massachusetts (D.A.R.)
| | - Benoit Lhermitte
- University of Alabama at Birmingham, Birmingham, Alabama (L.B.N.); Baylor University Medical Center, Dallas, Texas (K.L.F.); Henry Ford Hospital, Detroit, Michigan (T.M.); Clinic for Neurosurgery, Clinical Center of Serbia, Medical Faculty University of Belgrade, Belgrade, Serbia (D.G.); Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology Gliwice Branch, Radiotherapy and Chemotherapy Clinic, Gliwice, Poland (R.T.); Samsung Medical Center, Seoul, South Korea (D.H.N.); Centrum Onkologii Ziemi Lubelskiej, Lublin, Poland (M.M.); St. Luke's Brain Tumor Center, St. Luke's Hospital, Kansas City, Missouri (M.S.); Barrow Neurological Institute, Phoenix, Arizona (L.A.); Medical Oncology Unit 1, IOV, IRCCS, Padova, Italy (V.Z.); Policlinico di Modena, Modena, Italy (R.D.); Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (J.R.P.); Merck KGaA, Darmstadt, Germany (C.H., M.P.); Department of Clinical Neurosciences, University Hospital Lausanne, Lausanne, Switzerland (M.E.H.); Institute of Pathology, University Hospital Lausanne, Lausanne, Switzerland (B.L.); Dana-Farber Cancer Institute, Boston, Massachusetts (D.A.R.)
| | - David A Reardon
- University of Alabama at Birmingham, Birmingham, Alabama (L.B.N.); Baylor University Medical Center, Dallas, Texas (K.L.F.); Henry Ford Hospital, Detroit, Michigan (T.M.); Clinic for Neurosurgery, Clinical Center of Serbia, Medical Faculty University of Belgrade, Belgrade, Serbia (D.G.); Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology Gliwice Branch, Radiotherapy and Chemotherapy Clinic, Gliwice, Poland (R.T.); Samsung Medical Center, Seoul, South Korea (D.H.N.); Centrum Onkologii Ziemi Lubelskiej, Lublin, Poland (M.M.); St. Luke's Brain Tumor Center, St. Luke's Hospital, Kansas City, Missouri (M.S.); Barrow Neurological Institute, Phoenix, Arizona (L.A.); Medical Oncology Unit 1, IOV, IRCCS, Padova, Italy (V.Z.); Policlinico di Modena, Modena, Italy (R.D.); Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (J.R.P.); Merck KGaA, Darmstadt, Germany (C.H., M.P.); Department of Clinical Neurosciences, University Hospital Lausanne, Lausanne, Switzerland (M.E.H.); Institute of Pathology, University Hospital Lausanne, Lausanne, Switzerland (B.L.); Dana-Farber Cancer Institute, Boston, Massachusetts (D.A.R.)
| |
Collapse
|
53
|
Proportional upregulation of CD97 isoforms in glioblastoma and glioblastoma-derived brain tumor initiating cells. PLoS One 2015; 10:e0111532. [PMID: 25714433 PMCID: PMC4340952 DOI: 10.1371/journal.pone.0111532] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Accepted: 10/03/2014] [Indexed: 01/09/2023] Open
Abstract
CD97 is a novel glioma antigen that confers an invasive phenotype and poor survival in patients with glioblastoma (GBM), the most aggressive primary malignant brain tumor. The short isoform of CD97, known as EGF(1,2,5), has been shown to promote invasion and metastasis, but its role in gliomas and GBM-derived brain tumor initiating cells (BTICs) has not been studied. We sought to characterize CD97 expression among gliomas and identify the specific isoforms expressed. The short isoform of CD97 was identified in GBM and GBM-derived BTICs, but not low grade or anaplastic astrocytomas. All samples expressing the EGF(1,2,5) isoform were also found to express the EGF(1,2,3,5) isoform. These isoforms are believed to possess similar ligand binding patterns and interact with chondroitin sulfate, a component of the extracellular matrix, and the integrin α5β1. Using data acquired from the Cancer Genome Atlas (TCGA), we show that CD97 is upregulated among the classical and mesenchymal subtypes of GBM and significantly decreased among IDH1 mutant GBMs. Given its proven roles in tumor invasion, expression among aggressive genetic subtypes of GBM, and association with overall survival, CD97 is an attractive therapeutic target for patients with GBM.
Collapse
|
54
|
Rhun EL, Taillibert S, Chamberlain MC. The future of high-grade glioma: Where we are and where are we going. Surg Neurol Int 2015; 6:S9-S44. [PMID: 25722939 PMCID: PMC4338495 DOI: 10.4103/2152-7806.151331] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 10/15/2014] [Indexed: 01/12/2023] Open
Abstract
High-grade glioma (HGG) are optimally treated with maximum safe surgery, followed by radiotherapy (RT) and/or systemic chemotherapy (CT). Recently, the treatment of newly diagnosed anaplastic glioma (AG) has changed, particularly in patients with 1p19q codeleted tumors. Results of trials currenlty ongoing are likely to determine the best standard of care for patients with noncodeleted AG tumors. Trials in AG illustrate the importance of molecular characterization, which are germane to both prognosis and treatment. In contrast, efforts to improve the current standard of care of newly diagnosed glioblastoma (GB) with, for example, the addition of bevacizumab (BEV), have been largely disappointing and furthermore molecular characterization has not changed therapy except in elderly patients. Novel approaches, such as vaccine-based immunotherapy, for newly diagnosed GB are currently being pursued in multiple clinical trials. Recurrent disease, an event inevitable in nearly all patients with HGG, continues to be a challenge. Both recurrent GB and AG are managed in similar manner and when feasible re-resection is often suggested notwithstanding limited data to suggest benefit from repeat surgery. Occassional patients may be candidates for re-irradiation but again there is a paucity of data to commend this therapy and only a minority of selected patients are eligible for this approach. Consequently systemic therapy continues to be the most often utilized treatment in recurrent HGG. Choice of therapy, however, varies and revolves around re-challenge with temozolomide (TMZ), use of a nitrosourea (most often lomustine; CCNU) or BEV, the most frequently used angiogenic inhibitor. Nevertheless, no clear standard recommendation regarding the prefered agent or combination of agents is avaliable. Prognosis after progression of a HGG remains poor, with an unmet need to improve therapy.
Collapse
Affiliation(s)
- Emilie Le Rhun
- Department of Neuro-oncology, Roger Salengro Hospital, University Hospital, Lille, and Neurology, Department of Medical Oncology, Oscar Lambret Center, Lille, France, Inserm U-1192, Laboratoire de Protéomique, Réponse Inflammatoire, Spectrométrie de Masse (PRISM), Lille 1 University, Villeneuve D’Ascq, France
| | - Sophie Taillibert
- Neurology, Mazarin and Radiation Oncology, Pitié Salpétrière Hospital, University Pierre et Marie Curie, Paris VI, Paris, France
| | - Marc C. Chamberlain
- Department of Neurology and Neurological Surgery, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
55
|
Abstract
Despite decades of advancing science and clinical trials, average survival remains dismal for individuals with high-grade gliomas. Our understanding of the genetic and molecular aberrations that contribute to the aggressive nature of these tumors is continually growing, as is our ability to target such specific traits. Herein, we review the major classes of such targeted therapies, as well as the relevant clinical trial outcomes regarding their efficacy.
Collapse
Affiliation(s)
- Justin T Jordan
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, 450 Brookline Avenue, Boston, MA, 02215, USA
| | | |
Collapse
|
56
|
|
57
|
|
58
|
Iagaru A, Mosci C, Shen B, Chin FT, Mittra E, Telli ML, Gambhir SS. 18F-FPPRGD2 PET/CT: Pilot Phase Evaluation of Breast Cancer Patients. Radiology 2014; 273:549-59. [DOI: 10.1148/radiol.14140028] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
59
|
Abstract
The survival outcome of patients with malignant gliomas is still poor, despite advances in surgical techniques, radiation therapy and the development of novel chemotherapeutic agents. The heterogeneity of molecular alterations in signaling pathways involved in the pathogenesis of these tumors contributes significantly to their resistance to treatment. Several molecular targets for therapy have been discovered over the last several years. Therapeutic agents targeting these signaling pathways may provide more effective treatments and may improve survival. This review summarizes the important molecular therapeutic targets and the outcome of published clinical trials involving targeted therapeutic agents in glioma patients.
Collapse
|
60
|
Hottinger AF, Stupp R, Homicsko K. Standards of care and novel approaches in the management of glioblastoma multiforme. CHINESE JOURNAL OF CANCER 2014; 33:32-9. [PMID: 24384238 PMCID: PMC3905088 DOI: 10.5732/cjc.013.10207] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Glioblastoma multiforme (GBM) is the most common malignant primary brain tumor in adults. Standard therapeutic approaches provide modest improvement in the progression-free and overall survival, necessitating the investigation of novel therapies. We review the standard treatment options for GBM and evaluate the results obtained in clinical trials for promising novel approaches, including the inhibition of angiogenesis, targeted approaches against molecular pathways, immunotherapies, and local treatment with low voltage electric fields.
Collapse
Affiliation(s)
- Andreas F Hottinger
- Department of Clinical Neuroscience, Lausanne University Hospital, Lausanne 1011, Switzerland.
| | | | | |
Collapse
|
61
|
Whitson RJ, Lucia MS, Lambert JR. Growth differentiation factor-15 (GDF-15) suppresses in vitro angiogenesis through a novel interaction with connective tissue growth factor (CCN2). J Cell Biochem 2014; 114:1424-33. [PMID: 23280549 DOI: 10.1002/jcb.24484] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Accepted: 12/11/2012] [Indexed: 12/15/2022]
Abstract
Growth differentiation factor-15 (GDF-15) and the CCN family member, connective tissue growth factor (CCN2), are associated with cardiac disease, inflammation, and cancer. The precise role and signaling mechanism for these factors in normal and diseased tissues remains elusive. Here we demonstrate an interaction between GDF-15 and CCN2 using yeast two-hybrid assays and have mapped the domain of interaction to the von Willebrand factor type C domain of CCN2. Biochemical pull down assays using secreted GDF-15 and His-tagged CCN2 produced in PC-3 prostate cancer cells confirmed a direct interaction between these proteins. To investigate the functional consequences of this interaction, in vitro angiogenesis assays were performed. We demonstrate that GDF-15 blocks CCN2-mediated tube formation in human umbilical vein endothelial (HUVEC) cells. To examine the molecular mechanism whereby GDF-15 inhibits CCN2-mediated angiogenesis, activation of αV β3 integrins and focal adhesion kinase (FAK) was examined. CCN2-mediated FAK activation was inhibited by GDF-15 and was accompanied by a decrease in αV β3 integrin clustering in HUVEC cells. These results demonstrate, for the first time, a novel signaling pathway for GDF-15 through interaction with the matricellular signaling molecule CCN2. Furthermore, antagonism of CCN2 mediated angiogenesis by GDF-15 may provide insight into the functional role of GDF-15 in disease states.
Collapse
Affiliation(s)
- Ramon J Whitson
- Department of Pathology, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado 80045, USA
| | | | | |
Collapse
|
62
|
Stupp R, Hegi ME, Gorlia T, Erridge SC, Perry J, Hong YK, Aldape KD, Lhermitte B, Pietsch T, Grujicic D, Steinbach JP, Wick W, Tarnawski R, Nam DH, Hau P, Weyerbrock A, Taphoorn MJB, Shen CC, Rao N, Thurzo L, Herrlinger U, Gupta T, Kortmann RD, Adamska K, McBain C, Brandes AA, Tonn JC, Schnell O, Wiegel T, Kim CY, Nabors LB, Reardon DA, van den Bent MJ, Hicking C, Markivskyy A, Picard M, Weller M. Cilengitide combined with standard treatment for patients with newly diagnosed glioblastoma with methylated MGMT promoter (CENTRIC EORTC 26071-22072 study): a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol 2014; 15:1100-8. [PMID: 25163906 DOI: 10.1016/s1470-2045(14)70379-1] [Citation(s) in RCA: 729] [Impact Index Per Article: 72.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND Cilengitide is a selective αvβ3 and αvβ5 integrin inhibitor. Data from phase 2 trials suggest that it has antitumour activity as a single agent in recurrent glioblastoma and in combination with standard temozolomide chemoradiotherapy in newly diagnosed glioblastoma (particularly in tumours with methylated MGMT promoter). We aimed to assess cilengitide combined with temozolomide chemoradiotherapy in patients with newly diagnosed glioblastoma with methylated MGMT promoter. METHODS In this multicentre, open-label, phase 3 study, we investigated the efficacy of cilengitide in patients from 146 study sites in 25 countries. Eligible patients (newly diagnosed, histologically proven supratentorial glioblastoma, methylated MGMT promoter, and age ≥18 years) were stratified for prognostic Radiation Therapy Oncology Group recursive partitioning analysis class and geographic region and centrally randomised in a 1:1 ratio with interactive voice response system to receive temozolomide chemoradiotherapy with cilengitide 2000 mg intravenously twice weekly (cilengitide group) or temozolomide chemoradiotherapy alone (control group). Patients and investigators were unmasked to treatment allocation. Maintenance temozolomide was given for up to six cycles, and cilengitide was given for up to 18 months or until disease progression or unacceptable toxic effects. The primary endpoint was overall survival. We analysed survival outcomes by intention to treat. This study is registered with ClinicalTrials.gov, number NCT00689221. FINDINGS Overall, 3471 patients were screened. Of these patients, 3060 had tumour MGMT status tested; 926 patients had a methylated MGMT promoter, and 545 were randomly assigned to the cilengitide (n=272) or control groups (n=273) between Oct 31, 2008, and May 12, 2011. Median overall survival was 26·3 months (95% CI 23·8-28·8) in the cilengitide group and 26·3 months (23·9-34·7) in the control group (hazard ratio 1·02, 95% CI 0·81-1·29, p=0·86). None of the predefined clinical subgroups showed a benefit from cilengitide. We noted no overall additional toxic effects with cilengitide treatment. The most commonly reported adverse events of grade 3 or worse in the safety population were lymphopenia (31 [12%] in the cilengitide group vs 26 [10%] in the control group), thrombocytopenia (28 [11%] vs 46 [18%]), neutropenia (19 [7%] vs 24 [9%]), leucopenia (18 [7%] vs 20 [8%]), and convulsion (14 [5%] vs 15 [6%]). INTERPRETATION The addition of cilengitide to temozolomide chemoradiotherapy did not improve outcomes; cilengitide will not be further developed as an anticancer drug. Nevertheless, integrins remain a potential treatment target for glioblastoma. FUNDING Merck KGaA, Darmstadt, Germany.
Collapse
Affiliation(s)
- Roger Stupp
- UniversitätsSpital Zürich, Zurich, Switzerland; Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland.
| | - Monika E Hegi
- Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | | | - Sara C Erridge
- Edinburgh Cancer Centre, University of Edinburgh, Edinburgh, UK
| | - James Perry
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Yong-Kil Hong
- The Catholic University of Korea, Seoul St Mary's Hospital, Seoul, South Korea
| | - Kenneth D Aldape
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Benoit Lhermitte
- Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Torsten Pietsch
- Department of Neuropathology, Universität Bonn, Bonn, Germany
| | - Danica Grujicic
- Clinic for Neurosurgery, Clinical Center Serbia and Medical Faculty University of Belgrade, Belgrade, Serbia
| | | | - Wolfgang Wick
- Heidelberg University Medical Center & German Cancer Research Center, Heidelberg, Germany
| | - Rafał Tarnawski
- Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology Gliwice Branch, Gliwice, Poland
| | - Do-Hyun Nam
- Samsung Medical Center, Sungkyunkwan Univ School of Medicine, Seoul, South Korea
| | - Peter Hau
- Universitätsklinikum Regensburg, Regensburg, Germany
| | | | | | | | - Nalini Rao
- Bangalore Institute of Oncology, Bangalore, India
| | | | | | | | | | | | | | - Alba A Brandes
- Bellaria-Maggiore Hospital, AUSL-IRCCS Institute of Neurological Sciences-Bologna, Italy
| | | | | | | | - Chae-Yong Kim
- Seoul National University Bundang Hospital, SNU College of Medicine, Seoul, South Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
63
|
Li D, Zhao X, Zhang L, Li F, Ji N, Gao Z, Wang J, Kang P, Liu Z, Shi J, Chen X, Zhu Z. (68)Ga-PRGD2 PET/CT in the evaluation of Glioma: a prospective study. Mol Pharm 2014; 11:3923-9. [PMID: 25093246 PMCID: PMC4224544 DOI: 10.1021/mp5003224] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
![]()
Integrin αvβ3 is overexpressed
in both neovasculature and glioma cells. We aimed to evaluate 68gallium-BNOTA-PRGD2 (68Ga-PRGD2) as a new reagent
for noninvasive integrin αvβ3 imaging
in glioma patients. With informed consent, 12 patients with suspicious
brain glioma, as diagnosed by enhanced magnetic resonance imaging
(MRI) scanning, were enrolled to undergo 68Ga-PRGD2 PET/CT
and 18F-FDG PET/CT scans before surgery. The preoperative
images were compared and correlated with the pathologically determined
WHO grade. Next, the expression of integrin αvβ3, CD34, and Ki-67 were determined
by immunohistochemical staining of the resected brain tumor tissue.
Our findings demonstrated that 68Ga-PRGD2 specifically
accumulated in the brain tumors that were rich of integrin αvβ3 and other neovasculature markers, but
not in the brain parenchyma other than the choroid plexus. Therefore, 68Ga-PRGD2 PET/CT was able to evaluate the glioma demarcation
more specifically than 18F-FDG PET/CT. The maximum standardized
uptake values (SUVmax) of 68Ga-PRGD2, rather than those
of 18F-FDG, were significantly correlated with the glioma
grading. The maximum tumor-to-brain ratios (TBRmax) of both tracers
were significantly correlated with glioma grading, whereas 68Ga-PRGD2 seemed to be more superior to 18F-FDG in differentiating
high-grade glioma (HGG) from low-grade glioma (LGG). Moreover, 68Ga-PRGD2 PET/CT showed different accumulation patterns for
HGG of WHO grades III and IV. This is the first noninvasive integrin
imaging study, to the best of our knowledge, conducted in preoperative
patients with different grades of glioma, and it preliminarily indicated
the effectiveness of this novel method for evaluating glioma grading
and demarcation.
Collapse
Affiliation(s)
- Deling Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University ; China National Clinical Research Center for Neurological Diseases (NCRC-ND); Beijing Key Laboratory of Brian Tumor, Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
64
|
The role of targeted therapies in the management of progressive glioblastoma. J Neurooncol 2014; 118:557-99. [DOI: 10.1007/s11060-013-1339-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2013] [Accepted: 12/28/2013] [Indexed: 12/28/2022]
|
65
|
Heiduschka G, Lill C, Schneider S, Seemann R, Kornek G, Schmid R, Kotowski U, Thurnher D. The effect of cilengitide in combination with irradiation and chemotherapy in head and neck squamous cell carcinoma cell lines. Strahlenther Onkol 2014; 190:472-9. [PMID: 24557056 DOI: 10.1007/s00066-014-0600-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Accepted: 11/08/2013] [Indexed: 12/26/2022]
Abstract
BACKGROUND Integrins are highly attractive targets in oncology due to their involvement in angiogenesis in a wide spectrum of cancer entities. Among several integrin inhibitors under clinical evaluation, cilengitide is the most promising compound. However, little is known about the cellular processes induced during cilengitide therapy in combination with irradiation and cisplatin in head and neck squamous cell carcinoma (HNSCC). MATERIALS AND METHODS The cytostatic effect of cilengitide was assessed by proliferation assay in the three HNSCC cell lines SCC25, FaDu and CAL27. Combination experiments with cisplatin and irradiation were performed. Possible synergistic effects were calculated in combination index (CI) analyses. Colony forming inhibition was investigated in clonogenic assays. Real-time PCR arrays were used to evaluate target protein gene expression patterns. Flow cytometry was used to detect apoptosis. RESULTS Used alone, cilengitide has only minor cytotoxic effects in HNSCC cell lines. However, combination with cisplatin resulted in synergistic growth inhibition in all three cell lines. Irradiation showed synergism in short-term experiments and in colony forming assays, an additive effect was detected. Real-time PCR assay detected downregulation of the antiapoptotic protein Bcl-2 after exposure of cells to cilengitide. CONCLUSION Cilengitide in combination with cisplatin and irradiation may be a feasible option for the treatment of patients with head and neck cancer. However, further investigations are required to understand the exact mechanism that leads to synergistic cytotoxicity.
Collapse
Affiliation(s)
- G Heiduschka
- Department of Otorhinolaryngology, Head and Neck Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
66
|
Abstract
BACKGROUND Glioblastoma is the most common primary brain tumour. It has a poor prognosis despite some advances in treatment that have been achieved over the last ten years. In Slovenia, 50 to 60 glioblastoma patients are diagnosed each year. In order to establish whether the current treatment options have any influence on the survival of the Slovenian glioblastoma patients, their data in the period from the beginning of the year 1997 to the end of the year 2008 have been analysed. PATIENTS AND METHODS All patients treated at the Institute of Oncology Ljubljana from 1997 to 2008 were included in the retrospective study. Demographics, treatment details, and survival time after the diagnosis were collected and statistically analysed for the group as a whole and for subgroups. RESULTS From 1997 to 2008, 527 adult patients were diagnosed with glioblastoma and referred to the Institute of Oncology for further treatment. Their median age was 59 years (from 20 to 85) and all but one had the diagnosis confirmed by a pathologist. Gross total resection was reported by surgeons in 261 (49.5%) patients; good functional status (WHO 0 or 1) after surgery was observed in 336 (63.7%) patients, radiotherapy was performed in 422 (80.1%) patients, in 317 (75.1%) of them with radical intent, and 198 (62.5 %) of those received some form of systemic treatment (usually temozolomide). The median survival of all patients amounted to 9.7 months. There was no difference in median survival of all patients or of all treated patients before or after the chemo-radiotherapy era. However, the overall survival of patients treated with radical intent was significantly better (11.4 months; p < 0.05). A better survival was also noticed in radically treated patients who received additional temozolomide therapy (11.4 vs. 13.1 months; p = 0.014). The longer survival was associated with a younger age and a good performance status as well as with a more extensive tumour resection. In patients treated with radical intent, having a good performance status, and receiving radiotherapy and additional temozolomide therapy, the survival was significantly longer, based on multivariate analysis. CONCLUSIONS We observed a gradual increase in the survival of glioblastoma patients who were treated with radical intent over the last ten years. Good functional surgery, advances in radiotherapy and addition of temozolomide all contributed to this increase. Though the increased survival seems to be more pronounced in certain subgroups, we have still not been able to exactly define them. Further research, especially in tumour biology and genetics is needed.
Collapse
|
67
|
A conceptually new treatment approach for relapsed glioblastoma: coordinated undermining of survival paths with nine repurposed drugs (CUSP9) by the International Initiative for Accelerated Improvement of Glioblastoma Care. Oncotarget 2013; 4:502-30. [PMID: 23594434 PMCID: PMC3720600 DOI: 10.18632/oncotarget.969] [Citation(s) in RCA: 140] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
To improve prognosis in recurrent glioblastoma we developed a treatment protocol based on a combination of drugs not traditionally thought of as cytotoxic chemotherapy agents but that have a robust history of being well-tolerated and are already marketed and used for other non-cancer indications. Focus was on adding drugs which met these criteria: a) were pharmacologically well characterized, b) had low likelihood of adding to patient side effect burden, c) had evidence for interfering with a recognized, well-characterized growth promoting element of glioblastoma, and d) were coordinated, as an ensemble had reasonable likelihood of concerted activity against key biological features of glioblastoma growth. We found nine drugs meeting these criteria and propose adding them to continuous low dose temozolomide, a currently accepted treatment for relapsed glioblastoma, in patients with recurrent disease after primary treatment with the Stupp Protocol. The nine adjuvant drug regimen, Coordinated Undermining of Survival Paths, CUSP9, then are aprepitant, artesunate, auranofin, captopril, copper gluconate, disulfiram, ketoconazole, nelfinavir, sertraline, to be added to continuous low dose temozolomide. We discuss each drug in turn and the specific rationale for use- how each drug is expected to retard glioblastoma growth and undermine glioblastoma's compensatory mechanisms engaged during temozolomide treatment. The risks of pharmacological interactions and why we believe this drug mix will increase both quality of life and overall survival are reviewed.
Collapse
|
68
|
Abstract
Patients with glioblastoma typically present when tumors are at an advanced stage. Surgical resection, radiotherapy and adjuvant chemotherapy are currently the standard of care for glioblastoma. However, due to the infiltrative and dispersive nature of the tumor, recurrence rate remains high and typically results in very poor prognosis. Efforts to treat the primary tumor are, therefore, palliative rather than curative. From a practical perspective, controlling growth and dispersal of the recurrence may have a greater impact on disease-free survival. In order for cells to disperse, they must first detach from the mass. Preventing detachment may keep tumors that recur more localized and perhaps more amenable to therapy. Here we introduce a new perspective in which a quantifiable mechanical property, namely tissue surface tension, can provide novel information on tumor behavior. The overall theme of the discussion will attempt to integrate how adhesion molecules can alter a tumor's mechanical properties and how, in turn, these properties can be modified to prevent tumor cell detachment and dispersal.
Collapse
Affiliation(s)
- Ramsey A Foty
- Department of Surgery, University of Medicine & Dentistry, New Jersey Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA.
| |
Collapse
|
69
|
Onishi M, Kurozumi K, Ichikawa T, Date I. Mechanisms of tumor development and anti-angiogenic therapy in glioblastoma multiforme. Neurol Med Chir (Tokyo) 2013; 53:755-63. [PMID: 24162241 PMCID: PMC4508716 DOI: 10.2176/nmc.ra2013-0200] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Despite advances in surgical and medical therapy, glioblastoma multiforme (GBM) remains a fatal disease. There has been no significant increase in survival for patients with this disease over the last 20 years. Tumor vasculature formation and glioma cell invasion along the white matter tracts both play a pivotal role in glioma development. Angiogenesis and invasion are the major factors believed to be responsible for treatment resistance in tumors, and a better understanding of the glioma invasion and angiogenesis mechanisms will lead to the development of potential new treatments. In this review, we focus on the molecular characteristics of angiogenesis and invasion in human malignant glioma. We discuss bevacizumab and cilengitide, which are used to inhibit angiogenesis in GBM.
Collapse
Affiliation(s)
- Manabu Onishi
- Department of Neurological Surgery, Okayama, University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Okayama
- Address reprint requests to: Manabu Onishi, MD, PhD, Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama, Okayama 700-8558, Japan. e-mail:
| | - Kazuhiko Kurozumi
- Department of Neurological Surgery, Okayama, University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Okayama
| | - Tomotsugu Ichikawa
- Department of Neurological Surgery, Okayama, University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Okayama
| | - Isao Date
- Department of Neurological Surgery, Okayama, University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Okayama
| |
Collapse
|
70
|
MacDonald TJ, Vezina G, Stewart CF, Turner D, Pierson CR, Chen L, Pollack IF, Gajjar A, Kieran MW. Phase II study of cilengitide in the treatment of refractory or relapsed high-grade gliomas in children: a report from the Children's Oncology Group. Neuro Oncol 2013; 15:1438-44. [PMID: 24014381 DOI: 10.1093/neuonc/not058] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Cilengitide, an αv integrin antagonist, has demonstrated activity in recurrent adult glioblastoma (GBM). The Children's Oncology Group ACNS0621 study thus evaluated whether cilengitide is active as a single agent in the treatment of children with refractory high-grade glioma (HGG). Secondary objectives were to investigate the pharmacokinetics and pharmacogenomics of cilengitide in this population. METHODS Cilengitide (1800 mg/m(2)/dose intravenous) was administered twice weekly until evidence of disease progression or unacceptable toxicity. Thirty patients (age range, 1.1-20.3 years) were enrolled, of whom 24 were evaluable for the primary response end point. RESULTS Toxicity was infrequent and mild, with the exception of one episode of grade 2 pain possibly related to cilengitide. Two intratumoral hemorrhages were reported, but only one (grade 2) was deemed to be possibly related to cilengitide and was in the context of disease progression. One patient with GBM received cilengitide for 20 months and remains alive with continuous stable disease. There were no other responders, with median time to tumor progression of 28 days (range, 11-114 days). Twenty-one of the 24 evaluable patients died, with a median time from enrollment to death of 172 days (range, 28-325 days). The 3 patients alive at the time of this report had a follow-up time of 37, 223, and 1068 days, respectively. CONCLUSIONS We conclude that cilengitide is not effective as a single agent for refractory pediatric HGG. However, further study evaluating combination therapy with cilengitide is warranted before a role for cilengitide in the treatment of pediatric HGG can be excluded.
Collapse
Affiliation(s)
- Tobey J MacDonald
- Corresponding Author: Tobey J. MacDonald, MD, Emory Children's Center, Aflac Cancer and Blood Disorders Center, 2015 Uppergate Drive NE, Suite 442, Atlanta, GA 30322.
| | | | | | | | | | | | | | | | | |
Collapse
|
71
|
Kim YS, Li F, Kong R, Bai Y, Li KCP, Fan Y, O'Neill BE, Li Z. Multivalency of non-peptide integrin αVβ3 antagonist slows tumor growth. Mol Pharm 2013; 10:3603-11. [PMID: 23961901 DOI: 10.1021/mp400096z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Multivalency is a powerful strategy for achieving high-affinity molecular binding of compounds to increase their therapeutic potency or imaging potential. In our study, multivalent non-peptide integrin αvβ3 antagonists (IA) were designed for antitumor therapy. Docking and molecular dynamics were employed to explore the binding modes of IA monomer, dimer, and trimer. In silico, one IA unit binds tightly in the active site with similar pose to native ligand RGD and other parts of dimer and trimer contribute extra binding affinities by interacting with residues in vicinity of the original site. In vitro studies demonstrated that increasing valency results in increasing antiproliferative and antiorganizational effects against endothelial cells (HUVECs), and a much weaker effect on melanoma B16F10 cells. The antitumor efficacies of the IA multivalent compounds were evaluated in subcutaneous B16F10 melanoma tumor-bearing mice. At 30 mg/kg dose, the mean masses of tumors harvested 18 days after inoculation were significantly reduced (p<10(-7)) by 36±9%, 49±8%, and 71±7% for the IA monomer, dimer, and trimer groups, relative to control. The importance of multivalency was demonstrated to be highly significant beyond the additive effect of the extra pharmacological sites (p=0.00011). These results suggest that the major target of these anti-αvβ3 compounds is the neovasculature rather than the cancer cells, and the success of a multivalent strategy depends on the details of the components and linker. This is the first integrin αvβ3 multivalent ligand showing clear enhancement in antitumor effectiveness.
Collapse
Affiliation(s)
- Yoo-Shin Kim
- Department of Translational Imaging,§Department of Systems Medicine & Bioengineering, The Methodist Hospital Research Institute, Weill Medical College of Cornell University , 6670 Bertner Avenue, Houston, Texas 77030, United States
| | | | | | | | | | | | | | | |
Collapse
|
72
|
Scaringi C, Enrici RM, Minniti G. Combining molecular targeted agents with radiation therapy for malignant gliomas. Onco Targets Ther 2013; 6:1079-95. [PMID: 23966794 PMCID: PMC3745290 DOI: 10.2147/ott.s48224] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The expansion in understanding the molecular biology that characterizes cancer cells has led to the rapid development of new agents to target important molecular pathways associated with aberrant activation or suppression of cellular signal transduction pathways involved in gliomagenesis, including epidermal growth factor receptor, vascular endothelial growth factor receptor, mammalian target of rapamycin, and integrins signaling pathways. The use of antiangiogenic agent bevacizumab, epidermal growth factor receptor tyrosine kinase inhibitors gefitinib and erlotinib, mammalian target of rapamycin inhibitors temsirolimus and everolimus, and integrin inhibitor cilengitide, in combination with radiation therapy, has been supported by encouraging preclinical data, resulting in a rapid translation into clinical trials. Currently, the majority of published clinical studies on the use of these agents in combination with radiation and cytotoxic therapies have shown only modest survival benefits at best. Tumor heterogeneity and genetic instability may, at least in part, explain the poor results observed with a single-target approach. Much remains to be learned regarding the optimal combination of targeted agents with conventional chemoradiation, including the use of multipathways-targeted therapies, the selection of patients who may benefit from combined treatments based on molecular biomarkers, and the verification of effective blockade of signaling pathways.
Collapse
Affiliation(s)
- Claudia Scaringi
- Department of Radiation Oncology, Sant'Andrea Hospital, University Sapienza, Rome, Italy
| | | | | |
Collapse
|
73
|
Bieńkowski M, Piaskowski S, Stoczyńska-Fidelus E, Szybka M, Banaszczyk M, Witusik-Perkowska M, Jesień-Lewandowicz E, Jaskólski DJ, Radomiak-Załuska A, Jesionek-Kupnicka D, Sikorska B, Papierz W, Rieske P, Liberski PP. Screening for EGFR amplifications with a novel method and their significance for the outcome of glioblastoma patients. PLoS One 2013; 8:e65444. [PMID: 23762372 PMCID: PMC3675194 DOI: 10.1371/journal.pone.0065444] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Accepted: 04/24/2013] [Indexed: 01/18/2023] Open
Abstract
Glioblastoma is a highly aggressive tumour of the central nervous system, characterised by poor prognosis irrespective of the applied treatment. The aim of our study was to analyse whether the molecular markers of glioblastoma (i.e. TP53 and IDH1 mutations, CDKN2A deletion, EGFR amplification, chromosome 7 polysomy and EGFRvIII expression) could be associated with distinct prognosis and/or response to the therapy. Moreover, we describe a method which allows for a reliable, as well as time- and cost-effective, screening for EGFR amplification and chromosome 7 polysomy with quantitative Real-Time PCR at DNA level. In the clinical data, only the patient’s age had prognostic significance (continuous: HR = 1.04; p<0.01). At the molecular level, EGFRvIII expression was associated with a better prognosis (HR = 0.37; p = 0.04). Intriguingly, EGFR amplification was associated with a worse outcome in younger patients (HR = 3.75; p<0.01) and in patients treated with radiotherapy (HR = 2.71; p = 0.03). We did not observe any difference between the patients with the amplification treated with radiotherapy and the patients without such a treatment. Next, EGFR amplification was related to a better prognosis in combination with the homozygous CDKN2A deletion (HR = 0.12; p = 0.01), but to a poorer prognosis in combination with chromosome 7 polysomy (HR = 14.88; p = 0.01). Importantly, the results emphasise the necessity to distinguish both mechanisms of the increased EGFR gene copy number (amplification and polysomy). To conclude, although the data presented here require validation in different groups of patients, they strongly advocate the consideration of the patient’s tumour molecular characteristics in the selection of the therapy.
Collapse
Affiliation(s)
- Michał Bieńkowski
- Department of Molecular Pathology and Neuropathology, Chair of Oncology, Medical University of Lodz, Lodz, Poland.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
74
|
Patil SA, Hosni-Ahmed A, Jones TS, Patil R, Pfeffer LM, Miller DD. Novel approaches to glioma drug design and drug screening. Expert Opin Drug Discov 2013; 8:1135-51. [PMID: 23738794 DOI: 10.1517/17460441.2013.807248] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Gliomas are considered the most malignant form of brain tumors, and ranked among the most aggressive human cancers. Despite advance standard therapy the prognosis for patients with gliomas remains poor. Chemotherapy has played an important role as an adjuvant in treating gliomas. The efficacy of the chemotherapeutic drug is limited due to poor drug delivery and the inherent chemo- and radio-resistance. Challenges of the brain cancer therapy in clinical settings are; i) to overcome the chemo- and radio-resistance, ii) to improve drug delivery to tumors and iii) the development of effective drug screening procedures. AREAS COVERED In this review, the authors discuss clinically important chemotherapeutic agents used for treating malignant gliomas along with novel drug design approaches. The authors, furthermore, discuss the in vitro and in vivo drug screening procedures for the development of novel drug candidates. EXPERT OPINION The development of novel and highly potent chemotherapeutic agents for both glioma and glioma stem cells (GSCs) is highly important for future brain cancer research. Thus, research efforts should be directed towards developing innovative molecularly targeted antiglioma agents in order to reduce the toxicity and drug resistance which are associated with current forms of therapy. Development of novel pre-clinical drug screening procedures is also very critical for the overall success of brain cancer therapies in clinical settings.
Collapse
Affiliation(s)
- Shivaputra A Patil
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, 847 Monroe Avenue, Room 327, 881 Madison, Room 435, Memphis, TN 38163, USA.
| | | | | | | | | | | |
Collapse
|
75
|
Kurozumi K, Ichikawa T, Onishi M, Fujii K, Date I. Cilengitide treatment for malignant glioma: current status and future direction. Neurol Med Chir (Tokyo) 2013; 52:539-47. [PMID: 22976135 DOI: 10.2176/nmc.52.539] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Malignant glioma is the most common primary brain tumor and accounts for the majority of diagnoses. Treatment has involved a combination of surgery, radiation, and chemotherapy, yet these modalities rarely extend the life of the patient to more than one year from diagnosis. Integrins are expressed in tumor cells and tumor endothelial cells, and are important in angiogenesis and invasion in glioma. αvβ3 and αvβ5 integrins regulate cell adhesion, and inhibitors of these integrins suppress tumor growth in certain pre-clinical models. Several integrin-targeted drugs are in clinical trials as potential compounds for the treatment of cancer. Among them, cilengitide is a novel integrin antagonist for the treatment of glioblastoma. The multimodal anti-glioma effects are based on its cytotoxic, anti-angiogenic, anti-invasive, and synergetic effects. Preclinical studies showed a promising synergy between cilengitide and radiochemotherapy in order to normalize tumor vasculature and attenuate tumor invasion. Cilengitide is currently being assessed in phase III trials for patients with glioblastoma multiforme and in phase II trials for other types of cancers, demonstrating promising therapeutic outcomes to date. The results of these and other clinical studies are expected with great hope and interest. A more clear understanding of the benefits and pitfalls of each approach can then lead to the design of strategies to derive maximal benefit from these therapies.
Collapse
Affiliation(s)
- Kazuhiko Kurozumi
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Japan.
| | | | | | | | | |
Collapse
|
76
|
Niibori-Nambu A, Midorikawa U, Mizuguchi S, Hide T, Nagai M, Komohara Y, Nagayama M, Hirayama M, Kobayashi D, Tsubota N, Takezaki T, Makino K, Nakamura H, Takeya M, Kuratsu J, Araki N. Glioma initiating cells form a differentiation niche via the induction of extracellular matrices and integrin αV. PLoS One 2013; 8:e59558. [PMID: 23704872 PMCID: PMC3660593 DOI: 10.1371/journal.pone.0059558] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 02/15/2013] [Indexed: 01/10/2023] Open
Abstract
Glioma initiating cells (GICs) are considered responsible for the therapeutic resistance and recurrence of malignant glioma. To clarify the molecular mechanism of GIC maintenance/differentiation, we established GIC clones having the potential to differentiate into malignant gliomas, and subjected to DNA microarray/iTRAQ based integrated proteomics. 21,857 mRNAs and 8,471 proteins were identified and integrated into a gene/protein expression analysis chart. Gene Ontology analysis revealed that the expression of cell adhesion molecules, including integrin subfamilies, such as α2 and αV, and extracellular matrices (ECMs), such as collagen IV (COL4), laminin α2 (LAMA2), and fibronectin 1 (FN), was significantly upregulated during serum-induced GIC differentiation. This differentiation process, accompanied by the upregulation of MAPK as well as glioma specific proteins in GICs, was dramatically accelerated in these ECM (especially FN)-coated dishes. Integrin αV blocking antibody and RGD peptide significantly suppressed early events in GIC differentiation, suggesting that the coupling of ECMs to integrin αV is necessary for GIC differentiation. In addition, the expression of integrin αV and its strong ligand FN was prominently increased in glioblastomas developed from mouse intracranial GIC xenografts. Interestingly, during the initial phase of GIC differentiation, the RGD treatment significantly inhibited GIC proliferation and raised their sensitivity against anti-cancer drug temozolomide (TMZ). We also found that combination treatments of TMZ and RGD inhibit glioma progression and lead the longer survival of mouse intracranial GIC xenograft model. These results indicate that GICs induce/secrete ECMs to develop microenvironments with serum factors, namely differentiation niches that further stimulate GIC differentiation and proliferation via the integrin recognition motif RGD. A combination of RGD treatment with TMZ could have the higher inhibitory potential against the glioma recurrence that may be regulated by the GICs in the differentiation niche. This study provides a new perspective for developing therapeutic strategies against the early onset of GIC-associated glioma.
Collapse
Affiliation(s)
- Akiko Niibori-Nambu
- Department of Tumor Genetics and Biology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto-city, Japan
| | - Uichi Midorikawa
- Department of Tumor Genetics and Biology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto-city, Japan
- Healthcare Systems Laboratories, Sharp Corp. Chiba-city, Japan
| | - Souhei Mizuguchi
- Department of Tumor Genetics and Biology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto-city, Japan
| | - Takuichiro Hide
- Department of Neurosurgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto-city, Japan
| | - Minako Nagai
- Department of Tumor Genetics and Biology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto-city, Japan
| | - Yoshihiro Komohara
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto-city, Japan
| | - Megumi Nagayama
- Department of Tumor Genetics and Biology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto-city, Japan
| | - Mio Hirayama
- Department of Tumor Genetics and Biology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto-city, Japan
| | - Daiki Kobayashi
- Department of Tumor Genetics and Biology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto-city, Japan
| | - Nobuyuki Tsubota
- Department of Tumor Genetics and Biology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto-city, Japan
- Department of Neurosurgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto-city, Japan
| | - Tatsuya Takezaki
- Department of Neurosurgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto-city, Japan
| | - Keishi Makino
- Department of Neurosurgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto-city, Japan
| | - Hideo Nakamura
- Department of Neurosurgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto-city, Japan
| | - Motohiro Takeya
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto-city, Japan
| | - Junichi Kuratsu
- Department of Neurosurgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto-city, Japan
| | - Norie Araki
- Department of Tumor Genetics and Biology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto-city, Japan
- * E-mail:
| |
Collapse
|
77
|
Overexpression of CD97 confers an invasive phenotype in glioblastoma cells and is associated with decreased survival of glioblastoma patients. PLoS One 2013; 8:e62765. [PMID: 23658650 PMCID: PMC3637305 DOI: 10.1371/journal.pone.0062765] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Accepted: 03/24/2013] [Indexed: 12/23/2022] Open
Abstract
Mechanisms of invasion in glioblastoma (GBM) relate to differential expression of proteins conferring increased motility and penetration of the extracellular matrix. CD97 is a member of the epidermal growth factor seven-span transmembrane family of adhesion G-protein coupled receptors. These proteins facilitate mobility of leukocytes into tissue. In this study we show that CD97 is expressed in glioma, has functional effects on invasion, and is associated with poor overall survival. Glioma cell lines and low passage primary cultures were analyzed. Functional significance was assessed by transient knockdown using siRNA targeting CD97 or a non-target control sequence. Invasion was assessed 48 hours after siRNA-mediated knockdown using a Matrigel-coated invasion chamber. Migration was quantified using a scratch assay over 12 hours. Proliferation was measured 24 and 48 hours after confirmed protein knockdown. GBM cell lines and primary cultures were found to express CD97. Knockdown of CD97 decreased invasion and migration in GBM cell lines, with no difference in proliferation. Gene-expression based Kaplan-Meier analysis was performed using The Cancer Genome Atlas, demonstrating an inverse relationship between CD97 expression and survival. GBMs expressing high levels of CD97 were associated with decreased survival compared to those with low CD97 (p = 0.007). CD97 promotes invasion and migration in GBM, but has no effect on tumor proliferation. This phenotype may explain the discrepancy in survival between high and low CD97-expressing tumors. This data provides impetus for further studies to determine its viability as a therapeutic target in the treatment of GBM.
Collapse
|
78
|
Kornienko A, Mathieu V, Rastogi SK, Lefranc F, Kiss R. Therapeutic Agents Triggering Nonapoptotic Cancer Cell Death. J Med Chem 2013; 56:4823-39. [DOI: 10.1021/jm400136m] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Alexander Kornienko
- Department of Chemistry and Biochemistry, Texas State University—San Marcos, San Marcos, Texas 78666,
United States
| | - Véronique Mathieu
- Laboratoire
de Toxicologie, Faculté de Pharmacie, Université Libre de Bruxelles (ULB), Campus de la Plaine, CP205/1,
Boulevard du Triomphe, Brussels, Belgium
| | - Shiva K. Rastogi
- Department of Chemistry and Biochemistry, Texas State University—San Marcos, San Marcos, Texas 78666,
United States
| | - Florence Lefranc
- Service de Neurochirurgie, Hôpital Erasme, ULB, 808 Route de Lennik, 1070 Brussels, Belgium
| | - Robert Kiss
- Laboratoire
de Toxicologie, Faculté de Pharmacie, Université Libre de Bruxelles (ULB), Campus de la Plaine, CP205/1,
Boulevard du Triomphe, Brussels, Belgium
| |
Collapse
|
79
|
Fusicoccin a, a phytotoxic carbotricyclic diterpene glucoside of fungal origin, reduces proliferation and invasion of glioblastoma cells by targeting multiple tyrosine kinases. Transl Oncol 2013; 6:112-23. [PMID: 23544164 DOI: 10.1593/tlo.12409] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 01/29/2013] [Accepted: 01/30/2013] [Indexed: 01/27/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a deadly cancer that possesses an intrinsic resistance to pro-apoptotic insults, such as conventional chemotherapy and radiotherapy, and diffusely invades the brain parenchyma, which renders it elusive to total surgical resection. We found that fusicoccin A, a fungal metabolite from Fusicoccum amygdali, decreased the proliferation and migration of human GBM cell lines in vitro, including several cell lines that exhibit varying degrees of resistance to pro-apoptotic stimuli. The data demonstrate that fusicoccin A inhibits GBM cell proliferation by decreasing growth rates and increasing the duration of cell division and also decreases two-dimensional (measured by quantitative video microscopy) and three-dimensional (measured by Boyden chamber assays) migration. These effects of fusicoccin A treatment translated into structural changes in actin cytoskeletal organization and a loss of GBM cell adhesion. Therefore, fusicoccin A exerts cytostatic effects but low cytotoxic effects (as demonstrated by flow cytometry). These cytostatic effects can partly be explained by the fact that fusicoccin inhibits the activities of a dozen kinases, including focal adhesion kinase (FAK), that have been implicated in cell proliferation and migration. Overexpression of FAK, a nonreceptor protein tyrosine kinase, directly correlates with the invasive phenotype of aggressive human gliomas because FAK promotes cell proliferation and migration. Fusicoccin A led to the down-regulation of FAK tyrosine phosphorylation, which occurred in both normoxic and hypoxic GBM cell culture conditions. In conclusion, the current study identifies a novel compound that could be used as a chemical template for generating cytostatic compounds designed to combat GBM.
Collapse
|
80
|
Javerzat S, Godard V, Bikfalvi A. Balancing risks and benefits of anti-angiogenic drugs for malignant glioma. FUTURE NEUROLOGY 2013. [DOI: 10.2217/fnl.12.91] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Angiogenesis is a delicate process that has been programmed over the time of evolution of vertebrates to provide optimized quantities of oxygen and nutrients to the developing embryo and the growing newborn. Similarly, angiogenesis induction pathways are used during tumor development. Angiogenesis and tumor cell invasion are closely linked. Anti-angiogenesis treatment strategies have entered the clinic and show some promising results. However, recent research using preclinical models have pointed to possible harmful effects, including evasive resistance and increase in tumor cell invasion when VEGF activity is inhibited. This has been corroborated by observations in treated glioblastoma patients. However, the meaning of these observations is still in question. The results of Phase III clinical trials that are ongoing will certainly provide more definitive answers with regard to evasive resistance in glioblastoma treated with anti-angiogenic drugs.
Collapse
Affiliation(s)
- Sophie Javerzat
- University of Bordeaux, Laboratoire de l’Angiogenèse et du Microenvironnement des Cancers, Unités Mixte de Recherche 1029, F-33400 Talence, France
- Institut National de la Santé et de la Recherche Médicale, Laboratoire de l’Angiogenèse et du Microenvironnement des Cancers, Unités Mixte de Recherche 1029, F-33400 Talence, France
| | - Virginie Godard
- University of Bordeaux, Laboratoire de l’Angiogenèse et du Microenvironnement des Cancers, Unités Mixte de Recherche 1029, F-33400 Talence, France
- Institut National de la Santé et de la Recherche Médicale, Laboratoire de l’Angiogenèse et du Microenvironnement des Cancers, Unités Mixte de Recherche 1029, F-33400 Talence, France
| | - Andreas Bikfalvi
- Institut National de la Santé et de la Recherche Médicale, Laboratoire de l’Angiogenèse et du Microenvironnement des Cancers, Unités Mixte de Recherche 1029, F-33400 Talence, France
- University of Bordeaux, Laboratoire de l’Angiogenèse et du Microenvironnement des Cancers, Unités Mixte de Recherche 1029, F-33400 Talence, France.
| |
Collapse
|
81
|
Long-term response in high-grade optic glioma treated with medically induced hypothyroidism and carboplatin. Anticancer Drugs 2013; 24:315-23. [DOI: 10.1097/cad.0b013e32835c7a47] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
82
|
Inflammatory neovascularization during graft-versus-host disease is regulated by αv integrin and miR-100. Blood 2013; 121:3307-18. [PMID: 23327924 DOI: 10.1182/blood-2012-07-442665] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Acute graft-versus-host disease (GvHD) is a complex process involving endothelial damage and neovascularization. Better understanding of the pathophysiology of neovascularization during GvHD could help to target this process while leaving T-cell function intact. Under ischemic conditions, neovascularization is regulated by different micro RNAs (miRs), which potentially play a role in inflamed hypoxic GvHD target organs. We observed strong neovascularization in the murine inflamed intestinal tract (IT) during GvHD. Positron emission tomography imaging demonstrated abundant αvβ3 integrin expression within intestinal neovascularization areas. To interfere with neovascularization, we targeted αv integrin-expressing endothelial cells, which blocked their accumulation in the IT and reduced GvHD severity independent of immune reconstitution and graft-versus-tumor effects. Additionally, enhanced neovascularization and αv integrin expression correlated with GvHD severity in humans. Expression analysis of miRs in the inflamed IT of mice developing GvHD identified miR-100 as significantly downregulated. Inactivation of miR-100 enhanced GvHD indicating a protective role for miR-100 via blocking inflammatory neovascularization. Our data from the mouse model and patients indicate that inflammatory neovascularization is a central event during intestinal GvHD that can be inhibited by targeting αv integrin. We identify negative regulation of GvHD-related neovascularization by miR-100, which indicates common pathomechanistic features of GvHD and ischemia.
Collapse
|
83
|
Signaling determinants of glioma cell invasion. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 986:121-41. [PMID: 22879067 DOI: 10.1007/978-94-007-4719-7_7] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Tumor cell invasiveness is a critical challenge in the clinical management of glioma patients. In addition, there is accumulating evidence that current therapeutic modalities, including anti-angiogenic therapy and radiotherapy, can enhance glioma invasiveness. Glioma cell invasion is stimulated by both autocrine and paracrine factors that act on a large array of cell surface-bound receptors. Key signaling elements that mediate receptor-initiated signaling in the regulation of glioblastoma invasion are Rho family GTPases, including Rac, RhoA and Cdc42. These GTPases regulate cell morphology and actin dynamics and stimulate cell squeezing through the narrow extracellular spaces that are typical of the brain parenchyma. Transient attachment of cells to the extracellular matrix is also necessary for glioblastoma cell invasion. Interactions with extracellular matrix components are mediated by integrins that initiate diverse intracellular signalling pathways. Key signaling elements stimulated by integrins include PI3K, Akt, mTOR and MAP kinases. In order to detach from the tumor mass, glioma cells secrete proteolytic enzymes that cleave cell surface adhesion molecules, including CD44 and L1. Key proteases produced by glioma cells include uPA, ADAMs and MMPs. Increased understanding of the molecular mechanisms that control glioma cell invasion has led to the identification of molecular targets for therapeutic intervention in this devastating disease.
Collapse
|
84
|
Weller M, Cloughesy T, Perry JR, Wick W. Standards of care for treatment of recurrent glioblastoma--are we there yet? Neuro Oncol 2013; 15:4-27. [PMID: 23136223 PMCID: PMC3534423 DOI: 10.1093/neuonc/nos273] [Citation(s) in RCA: 530] [Impact Index Per Article: 48.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Accepted: 09/17/2012] [Indexed: 12/21/2022] Open
Abstract
Newly diagnosed glioblastoma is now commonly treated with surgery, if feasible, or biopsy, followed by radiation plus concomitant and adjuvant temozolomide. The treatment of recurrent glioblastoma continues to be a moving target as new therapeutic principles enrich the standards of care for newly diagnosed disease. We reviewed PubMed and American Society of Clinical Oncology abstracts from January 2006 to January 2012 to identify clinical trials investigating the treatment of recurrent or progressive glioblastoma with nitrosoureas, temozolomide, bevacizumab, and/or combinations of these agents. At recurrence, a minority of patients are eligible for second surgery or reirradiation, based on appropriate patient selection. In temozolomide-pretreated patients, progression-free survival rates at 6 months of 20%-30% may be achieved either with nitrosoureas, temozolomide in various dosing regimens, or bevacizumab. Combination regimens among these agents or with other drugs have not produced evidence for superior activity but commonly produce more toxicity. More research is needed to better define patient profiles that predict benefit from the limited therapeutic options available after the current standard of care has failed.
Collapse
Affiliation(s)
- Michael Weller
- Department of Neurology, University Hospital Zurich, Frauenklinikstrasse 26, CH-8091 Zurich, Switzerland.
| | | | | | | |
Collapse
|
85
|
Flexible or fixed: a comparative review of linear and cyclic cancer-targeting peptides. Future Med Chem 2012; 4:1601-18. [PMID: 22917248 DOI: 10.4155/fmc.12.75] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Peptides can serve as versatile cancer-targeting ligands and have been used for clinically relevant applications such as cancer imaging and therapy. A current and long-standing focus within peptide research is the creation of structurally constrained peptides generated through cyclization. Cyclization is envisioned to enhance the selective binding, uptake, potency and stability of linear precursors. This review compares closely related linear and cyclic peptides in these respects. Peptide cyclization generally improves the selective binding and stability of linear precursors; however, not all cyclization strategies and constrained geometries enhance these properties to the same extent. In some instances, linear analogues actually have better cancer-targeting properties compared with their cyclic counterparts. Although cyclization does not necessarily improve the cancer-targeting properties of linear analogues, cyclic peptides may obtain properties that allow them to be used for additional applications. This review aims to convey the advantages and limitations of cyclic cancer-targeting peptides.
Collapse
|
86
|
Boohaker RJ, Lee MW, Vishnubhotla P, Perez JM, Khaled AR. The use of therapeutic peptides to target and to kill cancer cells. Curr Med Chem 2012; 19:3794-804. [PMID: 22725698 DOI: 10.2174/092986712801661004] [Citation(s) in RCA: 226] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2011] [Revised: 04/27/2012] [Accepted: 05/03/2012] [Indexed: 02/06/2023]
Abstract
Peptide therapeutics is a promising field for emerging anti-cancer agents. Benefits include the ease and rapid synthesis of peptides and capacity for modifications. An existing and vast knowledge base of protein structure and function can be exploited for novel peptide design. Current research focuses on developing peptides that can (1) serve as tumor targeting moieties and (2) permeabilize membranes with cytotoxic consequences. A survey of recent findings reveals significant trends. Amphiphilic peptides with clusters of hydrophobic and cationic residues are features of anti-microbial peptides that confer the ability to eradicate microbes and show considerable anti-cancer toxicity. Peptides that assemble and form pores can disrupt cell or organelle membranes and cause apoptotic or necrotic death. Cell permeable and tumor-homing peptides can carry biologically active cargo to tumors or tumor vasculature. The challenge lies in developing the clinical application of therapeutic peptides. Improving delivery to tumors, minimizing non-specific toxic effects and discerning pharmacokinetic properties are high among the needs to produce a powerful therapeutic peptide for cancer treatment.
Collapse
Affiliation(s)
- R J Boohaker
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, 6900 Lake Nona Blvd, Orlando, FL 32827, USA
| | | | | | | | | |
Collapse
|
87
|
Cilengitide in bevacizumab-refractory high-grade glioma: two case reports and critical review of the literature. Anticancer Drugs 2012; 23:749-53. [PMID: 22382388 DOI: 10.1097/cad.0b013e3283520e2c] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
High-grade gliomas (HGG) are aggressive and highly vascularized brain tumours. Despite multimodality therapy including surgery, radiation therapy and in many cases temozolomide chemotherapy, the prognosis is dismal. Salvage therapies following progression after radiation therapy and chemotherapy have historically yielded disappointing results. Bevacizumab is an interesting antiangiogenic drug used as a second-line treatment but although most patients benefit, essentially all patients ultimately progress. Moreover, some clinical studies have documented low activity of a second attempt at vascular endothelial growth factor pathway inhibition after failure of a first. The use of another drug with a different angiogenic pathway inhibition may probably result in a higher activity. Here, we describe, to our knowledge for the first time, the activity and safety of cilengitide, an agent with a different antiangiogenic and anti-invasive activity, administered in two bevacizumab-refractory patients with HGG. In addition, we present a rapid review of the activity of cilengitide in HGG.
Collapse
|
88
|
Onishi M, Ichikawa T, Kurozumi K, Fujii K, Yoshida K, Inoue S, Michiue H, Chiocca EA, Kaur B, Date I. Bimodal anti-glioma mechanisms of cilengitide demonstrated by novel invasive glioma models. Neuropathology 2012; 33:162-74. [PMID: 22989076 DOI: 10.1111/j.1440-1789.2012.01344.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Revised: 08/07/2012] [Accepted: 08/09/2012] [Indexed: 12/12/2022]
Abstract
Integrins are expressed in tumor cells and tumor endothelial cells, and likely play important roles in glioma angiogenesis and invasion. We investigated the anti-glioma mechanisms of cilengitide (EMD121974), an αvβ3 integrin inhibitor, utilizing the novel invasive glioma models, J3T-1 and J3T-2. Immunohistochemical staining of cells in culture and brain tumors in rats revealed positive αvβ3 integrin expression in J3T-2 cells and tumor endothelial cells, but not in J3T-1 cells. Established J3T-1 and J3T-2 orthotopic gliomas in athymic rats were treated with cilengitide or solvent. J3T-1 gliomas showed perivascular tumor cluster formation and angiogenesis, while J3T-2 gliomas showed diffuse single-cell infiltration without obvious angiogenesis. Cilengitide treatment resulted in a significantly decreased diameter of the J3T-1 tumor vessel clusters and its core vessels when compared with controls, while an anti-invasive effect was shown in the J3T-2 glioma with a significant reduction of diffuse cell infiltration around the tumor center. The survival of cilengitide-treated mice harboring J3T-1 tumors was significantly longer than that of control animals (median survival: 57.5 days and 31.8 days, respectively, P < 0.005), while cilengitide had no effect on the survival of mice with J3T-2 tumors (median survival: 48.9 days and 48.5, P = 0.69). Our results indicate that cilengitide exerts a phenotypic anti-tumor effect by inhibiting angiogenesis and glioma cell invasion. These two mechanisms are clearly shown by the experimental treatment of two different animal invasive glioma models.
Collapse
Affiliation(s)
- Manabu Onishi
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Grimm SA, Chamberlain MC. State of the art and perspectives in the treatment of glioblastoma. CNS Oncol 2012; 1:49-70. [PMID: 25054300 PMCID: PMC6176827 DOI: 10.2217/cns.12.5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Glioblastoma is the most common malignant primary brain tumor. Cures are rare and median survival varies from several to 22 months. Standard treatment for good performance patients consists of maximal safe surgical resection followed by radiotherapy with concurrent temozolomide (TMZ) chemotherapy and six cycles of postradiotherapy TMZ. At recurrence, treatment options include repeat surgery (with or without Gliadel wafer placement), reirradiation or systemic therapy. Most patients with good performance status are treated with cytotoxic chemotherapy or targeted biologic therapy following or in lieu of repeat surgery. Cytotoxic chemotherapy options include nitrosoureas, rechallenge with TMZ, platins, phophoramides and topoisomerase inhibitors, although efficacy is limited. Despite the intense effort of developing biologic agents that target angiogenesis and growth and proliferative pathways, bevacizumab is the only agent that has shown efficacy in clinical trials. It was awarded accelerated approval in the USA after demonstrating an impressive radiographic response in two open-label, prospective Phase II studies. Two randomized, Phase III trials of upfront bevacizumab have completed and may demonstrate survival benefit; however, results are pending at this time. Given the limited treatment options at tumor recurrence, consideration for enrollment on a clinical trial is encouraged.
Collapse
Affiliation(s)
- Sean A Grimm
- Department of Neurology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Marc C Chamberlain
- Department of Neurology & Neurological Surgery, Seattle Cancer Care Alliance, Fred Hutchinson Cancer Research Center, University of Washington, 825 Eastlake Avenue E, PO Box 19023, MS-G4940, Seattle, WA 98109-1023, USA
| |
Collapse
|
90
|
Silber JR, Bobola MS, Blank A, Chamberlain MC. O(6)-methylguanine-DNA methyltransferase in glioma therapy: promise and problems. BIOCHIMICA ET BIOPHYSICA ACTA 2012; 1826:71-82. [PMID: 22244911 PMCID: PMC3340514 DOI: 10.1016/j.bbcan.2011.12.004] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Revised: 12/28/2011] [Accepted: 12/29/2011] [Indexed: 11/23/2022]
Abstract
Gliomas are the most frequent adult primary brain tumor, and are invariably fatal. The most common diagnosis glioblastoma multiforme (GBM) afflicts 12,500 new patients in the U.S. annually, and has a median survival of approximately one year when treated with the current standard of care. Alkylating agents have long been central in the chemotherapy of GBM and other gliomas. The DNA repair protein O(6)-methylguanine-DNA methyltransferase (MGMT), the principal human activity that removes cytotoxic O(6)-alkylguanine adducts from DNA, promotes resistance to anti-glioma alkylators, including temozolomide and BCNU, in GBM cell lines and xenografts. Moreover, MGMT expression assessed by immunohistochemistry, biochemical activity or promoter CpG methylation status is associated with the response of GBM to alkylator-based therapies, providing evidence that MGMT promotes clinical resistance to alkylating agents. These observations suggest a role for MGMT in directing adjuvant therapy of GBM and other gliomas. Promoter methylation status is the most clinically tractable measure of MGMT, and there is considerable enthusiasm for exploring its utility as a marker to assign therapy to individual patients. Here, we provide an overview of the biochemical, genetic and biological characteristics of MGMT as they relate to glioma therapy. We consider current methods to assess MGMT expression and discuss their utility as predictors of treatment response. Particular emphasis is given to promoter methylation status and the methodological and conceptual impediments that limit its use to direct treatment. We conclude by considering approaches that may improve the utility of MGMT methylation status in planning optimal therapies tailored to individual patients.
Collapse
Affiliation(s)
- John R Silber
- Department of Neurological Surgery, University of Washington, Seattle, WA 98195, USA.
| | | | | | | |
Collapse
|
91
|
Antiangiogenic therapy for glioma. JOURNAL OF SIGNAL TRANSDUCTION 2012; 2012:483040. [PMID: 22830012 PMCID: PMC3399341 DOI: 10.1155/2012/483040] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Revised: 04/27/2012] [Accepted: 05/02/2012] [Indexed: 01/18/2023]
Abstract
Currently, antiangiogenic agents are routinely used for the treatment of patients with glioma. However, despite advances in pharmacological and surgical therapy, glioma remains an incurable disease. Indeed, the formation of an abnormal tumor vasculature and the invasion of glioma cells along neuronal tracts are proposed to comprise the major factors that are attributed to the therapeutic resistance of these tumors. The development of curative therapeutic modalities for the treatment of glioma requires further investigation of the molecular mechanisms regulating angiogenesis and invasion. In this review, we discuss the molecular characteristics of angiogenesis and invasion in human malignant glioma, we present several available drugs that are used or can potentially be utilized for the inhibition of angiogenesis in glioma, and we focus our attention on the key mediators of the molecular mechanisms underlying the resistance of glioma to antiangiogenic therapy.
Collapse
|
92
|
Roxin Á, Chen J, Scully CCG, Rotstein BH, Yudin AK, Zheng G. Conformational Modulation of in Vitro Activity of Cyclic RGD Peptides via Aziridine Aldehyde-Driven Macrocyclization Chemistry. Bioconjug Chem 2012; 23:1387-95. [DOI: 10.1021/bc300239a] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Áron Roxin
- Ontario Cancer Institute and
Princess Margaret Hospital, University Health Network, Toronto, Ontario, Canada
| | - Juan Chen
- Ontario Cancer Institute and
Princess Margaret Hospital, University Health Network, Toronto, Ontario, Canada
| | | | | | | | - Gang Zheng
- Ontario Cancer Institute and
Princess Margaret Hospital, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
93
|
Focal adhesion-chromatin linkage controls tumor cell resistance to radio- and chemotherapy. CHEMOTHERAPY RESEARCH AND PRACTICE 2012; 2012:319287. [PMID: 22778951 PMCID: PMC3385588 DOI: 10.1155/2012/319287] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Revised: 04/17/2012] [Accepted: 05/07/2012] [Indexed: 11/29/2022]
Abstract
Cancer resistance to therapy presents an ongoing and unsolved obstacle, which has clear impact on patient's survival. In order to address this problem, novel in vitro models have been established and are currently developed that enable data generation in a more physiological context. For example, extracellular-matrix- (ECM-) based scaffolds lead to the identification of integrins and integrin-associated signaling molecules as key promoters of cancer cell resistance to radio- and chemotherapy as well as modern molecular agents. In this paper, we discuss the dynamic nature of the interplay between ECM, integrins, cytoskeleton, nuclear matrix, and chromatin organization and how this affects the response of tumor cells to various kinds of cytotoxic anticancer agents.
Collapse
|
94
|
Liu X, Cui W, Li B, Hong Z. Targeted therapy for glioma using cyclic RGD-entrapped polyionic complex nanomicelles. Int J Nanomedicine 2012; 7:2853-62. [PMID: 22745548 PMCID: PMC3383325 DOI: 10.2147/ijn.s29788] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND The purpose of this study was to test the efficacy of cyclic Arg-Gly-Asp (RGD) peptide conjugated with polyionic complex nanomicelles as targeted therapy for glioma. METHODS A stable cyclic RGD polyionic complex nanostructure, ie, a c(RGDfC) polyionic complex micelle, was synthesized and its biocompatibility with cultured neurons was assessed using a cell viability assay. Targeted binding to cultured glioma cells was evaluated by the CdTe quantum dot marking technique and a cell viability assay. The inhibitory effect of the nanomicelles against glioma cells was also evaluated, and their targeted migration into rat brain glioma cells and apoptotic effects were traced by the CdTe quantum dot marking and immunohistochemical staining. RESULTS c(RGDfC) polyionic complex micelles did not affect the growth of neurons but bonded selectively to and inhibited proliferation of glioma cells in vitro. When tested in vivo, the micelles migrated into glioma cells, inducing apoptosis in the rat brain. CONCLUSION The c(RGDfC) polyionic complex micelle is an effective targeted therapy against glioma.
Collapse
Affiliation(s)
- Xiaoying Liu
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai China.
| | | | | | | |
Collapse
|
95
|
Bianchini F, Cini N, Trabocchi A, Bottoncetti A, Raspanti S, Vanzi E, Menchi G, Guarna A, Pupi A, Calorini L. ¹²⁵I-radiolabeled morpholine-containing arginine-glycine-aspartate (RGD) ligand of αvβ₃ integrin as a molecular imaging probe for angiogenesis. J Med Chem 2012; 55:5024-33. [PMID: 22621422 DOI: 10.1021/jm2016232] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
In this paper, using a hybrid small-animal Micro SPECT/CT imaging system, we report that a new (125)I-Cilengitide-like RGD-cyclopentapeptide, containing d-morpholine-3-carboxylic acid, interacts in vivo with α(v)β(3) integrin expressed by melanoma cells. Images clearly show that the (125)I-compound has the capacity to monitor the growth of a melanoma xenograft. Indeed, retention of the labeled ligand in the tumor mass has a good tumor/background ratio, and a significant reduction of its uptake was observed after injection of unlabeled ligand. These results suggest that the use of (125)I-labeled morpholine-based RGD-cyclopentapeptides targeting α(v)β(3) positive tumors may play a role in future therapeutic strategies.
Collapse
Affiliation(s)
- Francesca Bianchini
- Department of Experimental Pathology and Oncology, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Anton K, Baehring JM, Mayer T. Glioblastoma multiforme: overview of current treatment and future perspectives. Hematol Oncol Clin North Am 2012; 26:825-53. [PMID: 22794286 DOI: 10.1016/j.hoc.2012.04.006] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Glioblastoma multiforme is the most common primary malignant tumor of the central nervous system. Despite new insights into glioblastoma pathophysiology, the prognosis for patients diagnosed with this highly aggressive tumor remains bleak. Current treatment regimens combine surgical resection and chemoradiotherapy, providing an increase in median overall survival from 12.1 to 14.6 months. Ongoing preclinical and clinical studies evaluating the efficacy of novel therapies provide hope for increasing survival benefit. This article reviews the advancements in glioblastoma treatment in newly diagnosed and recurrent glioblastoma, including novel therapies such as antiangiogenic agents, mammalian target of rapamycin inhibitors, poly(ADP-ribose) polymerase-1 inhibitors, and immunotherapies.
Collapse
Affiliation(s)
- Kevin Anton
- Department of Pharmacology, Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ 08903, USA
| | | | | |
Collapse
|
97
|
Svensen N, Walton JG, Bradley M. Peptides for cell-selective drug delivery. Trends Pharmacol Sci 2012; 33:186-92. [DOI: 10.1016/j.tips.2012.02.002] [Citation(s) in RCA: 127] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Revised: 02/14/2012] [Accepted: 02/14/2012] [Indexed: 10/28/2022]
|
98
|
Risk of ischemia in glioma surgery: comparison of first and repeat procedures. J Neurooncol 2012; 107:599-607. [DOI: 10.1007/s11060-011-0784-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Accepted: 12/26/2011] [Indexed: 10/14/2022]
|
99
|
Gilbert MR. Establishing the standard of care for patients with newly diagnosed and recurrent glioblastoma. Am Soc Clin Oncol Educ Book 2012:112-7. [PMID: 24451719 DOI: 10.14694/edbook_am.2012.32.197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The current standard of care for patients with newly diagnosed glioblastoma includes maximal safe tumor resection followed by concurrent external-beam radiation with daily low-dose temozolomide followed by 6 to 12 months of adjuvant temozolomide, typically by using a cycle of 5 consecutive days out of 28. Efforts to improve on these results from the European Organisation for Research and Treatment of Cancer (EORTC)/National Cancer Institute of Canada (NCIC) trial using either dose-dense chemotherapy strategies or combinations with signal transduction modulators have, to date, been unsuccessful. Two large international randomized trials examining the efficacy of adding bevacizumab, an antiangiogenic agent, to the standard treatment have been completed, with expectations of results within in the next 2 years. For recurrent glioblastoma, there are no firmly established standards of care. Although intracavitary insertion of carmustine-impregnated polymers has been approved by the U.S. Food and Drug Administration (FDA), this strategy is not widely used. Bevacizumab has been FDA approved for recurrent glioblastoma, but no randomized trial has clearly demonstrated a survival benefit. Alternative dosing schedules of temozolomide (i.e., metronomic) has modest activity even in patients with prior temozolomide exposure. Clinical trials testing small-molecule signal transduction modulators have been disappointing, although most report a small response rate, suggesting that molecularly definable tumor subpopulations may help guide treatment decisions. Successful implementation of marker-based treatment would lead to personalized care and the creation of individualized standards of care.
Collapse
Affiliation(s)
- Mark R Gilbert
- From the Department of Neuro-oncology, University of Texas M. D. Anderson Cancer Center, Houston, TX
| |
Collapse
|
100
|
Reardon DA, Cheresh D. Cilengitide: a prototypic integrin inhibitor for the treatment of glioblastoma and other malignancies. Genes Cancer 2011; 2:1159-65. [PMID: 22866207 PMCID: PMC3411133 DOI: 10.1177/1947601912450586] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Integrins are critical intermediaries in a wide spectrum of cancer cell activities and thus represent a highly attractive target in oncology therapy. Nonetheless, successful exploitation of anti-integrin therapeutics has proven challenging to date for cancer patients. In this review, we will focus on cilengitide, an RGD pentapeptide inhibitor of α V integrins. Although several integrin inhibitors are under clinical evaluation, cilengitide is the most clinically advanced and is emerging as a prototype for this class of anticancer therapy. A foundation of encouraging preclinical studies led to a well-designed clinical development plan that culminated in a pivotal phase III study of cilengitide in combination with radiation therapy and temozolomide chemotherapy for newly diagnosed glioblastoma patients. Accrual to this study recently completed, while phase II studies of cilengitide are ongoing for head and neck cancer as well as lung cancer. Important future considerations for cilengitide and other integrin-targeting agents will likely include the identification of optimal combinatorial regimens and the delineation of biomarkers associated with efficacy.
Collapse
Affiliation(s)
- David A. Reardon
- Department of Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - David Cheresh
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|